Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Feedback regulation between plasma membrane tension and membrane-bending proteins organizes cell polarity during leading edge formation

Abstract

Tension applied to the plasma membrane (PM) is a global mechanical parameter involved in cell migration. However, how membrane tension regulates actin assembly is unknown. Here, we demonstrate that FBP17, a membrane-bending protein and an activator of WASP/N-WASP-dependent actin nucleation, is a PM tension sensor involved in leading edge formation. In migrating cells, FBP17 localizes to short membrane invaginations at the leading edge, while diminishing from the cell rear in response to PM tension increase. Conversely, following reduced PM tension, FBP17 dots randomly distribute throughout the cell, correlating with loss of polarized actin assembly on PM tension reduction. Actin protrusive force is required for the polarized accumulation, indicating a role for FBP17-mediated activation of WASP/N-WASP in PM tension generation. In vitro experiments show that FBP17 membrane-bending activity depends on liposomal membrane tension. Thus, FBP17 is the local activator of actin polymerization that is inhibited by PM tension in the feedback loop that regulates cell migration.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Essential role for PM tension in polarized distribution of FBP17.
Figure 2: Polarization of GFP–FBP17 by PM tension increase.
Figure 3: FBP17 plays an important role in PM tension-dependent leading edge formation.
Figure 4: Membrane-bending ability is essential for polarized distribution of FBP17.
Figure 5: Membrane-bending activity of FBP17 depends on membrane tension.
Figure 6: Reciprocal interaction between membrane-bending activity and actin-based protrusive force is required for polarized distribution of FBP17.
Figure 7: Mechanism of leading edge formation by FBP17.

Similar content being viewed by others

References

  1. Jilkine, A. & Edelstein-Keshet, L. A comparison of mathematical models for polarization of single eukaryotic cells in response to guided cues. PLoS Comput. Biol. 7, e1001121 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Neilson, M. P. et al. Chemotaxis: a feedback-based computational model robustly predicts multiple aspects of real cell behaviour. PLoS Biol. 9, e1000618 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Graziano, B. R. & Weiner, O. D. Self-organization of protrusions and polarity during eukaryotic chemotaxis. Curr. Opin. Cell Biol. 30, 60–67 (2014).

    Article  CAS  PubMed  Google Scholar 

  4. Weiner, O. D., Marganski, W. A., Wu, L. F., Altschuler, S. J. & Kirschner, M. W. An actin-based wave generator organizes cell motility. PLoS Biol. 5, e221 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Veltman, D. M., King, J. S., Machesky, L. M. & Insall, R. H. SCAR knockouts in Dictyostelium: WASP assumes SCAR’s position and upstream regulators in pseudopods. J. Cell Biol. 198, 501–508 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Huang, C. H., Tang, M., Shi, C., Iglesias, P. A. & Devreotes, P. N. An excitable signal integrator couples to an idling cytoskeletal oscillator to drive cell migration. Nat. Cell Biol. 15, 1307–1316 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Allard, J. & Mogilner, A. Traveling waves in actin dynamics and cell motility. Curr. Opin. Cell Biol. 25, 107–115 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Iglesias, P. A. & Devreotes, P. N. Biased excitable networks: how cells direct motion in response to gradients. Curr. Opin. Cell Biol. 24, 245–253 (2012).

    Article  CAS  PubMed  Google Scholar 

  9. Houk, A. R. et al. Membrane tension maintains cell polarity by confining signals to the leading edge during neutrophil migration. Cell 148, 175–188 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Gauthier, N. C., Masters, T. A. & Sheetz, M. P. Mechanical feedback between membrane tension and dynamics. Trends Cell Biol. 22, 527–535 (2012).

    Article  CAS  PubMed  Google Scholar 

  11. Diz-Munoz, A., Fletcher, D. A. & Weiner, O. D. Use the force: membrane tension as an organizer of cell shape and motility. Trends Cell Biol. 23, 47–53 (2013).

    Article  CAS  PubMed  Google Scholar 

  12. Lieber, A. D., Yehudai-Resheff, S., Barnhart, E. L., Theriot, J. A. & Keren, K. Membrane tension in rapidly moving cells is determined by cytoskeletal forces. Curr. Biol. 23, 1409–1417 (2013).

    Article  CAS  PubMed  Google Scholar 

  13. Ho, H. Y. et al. Toca-1 mediates Cdc42-dependent actin nucleation by activating the N-WASP-WIP complex. Cell 118, 203–216 (2004).

    Article  CAS  PubMed  Google Scholar 

  14. Hu, J., Mukhopadhyay, A. & Craig, A. W. Transducer of Cdc42-dependent actin assembly promotes epidermal growth factor-induced cell motility and invasiveness. J. Biol. Chem. 286, 2261–2272 (2011).

    Article  CAS  PubMed  Google Scholar 

  15. Malet-Engra, G. et al. CIP4 controls CCL19-driven cell steering and chemotaxis in chronic lymphocytic leukemia. Cancer Res. 73, 3412–3424 (2013).

    Article  CAS  PubMed  Google Scholar 

  16. Chander, H., Truesdell, P., Meens, J. & Craig, A. W. Transducer of Cdc42-dependent actin assembly promotes breast cancer invasion and metastasis. Oncogene 32, 3080–3090 (2013).

    Article  CAS  PubMed  Google Scholar 

  17. Rolland, Y. et al. The CDC42-interacting protein 4 controls epithelial cell cohesion and tumor dissemination. Dev. Cell 30, 553–568 (2014).

    Article  CAS  PubMed  Google Scholar 

  18. Truesdell, P. et al. CIP4 promotes lung adenocarcinoma metastasis and is associated with poor prognosis. Oncogene (2014)10.1038/onc.2014.280

  19. Shimada, A. et al. Curved EFC/F-BAR-domain dimers are joined end to end into a filament for membrane invagination in endocytosis. Cell 129, 761–772 (2007).

    Article  CAS  PubMed  Google Scholar 

  20. Frost, A. et al. Structural basis of membrane invagination by F-BAR domains. Cell 132, 807–817 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Liu, J., Sun, Y., Drubin, D. G. & Oster, G. F. The mechanochemistry of endocytosis. PLoS Biol. 7, e1000204 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Takano, K., Toyooka, K. & Suetsugu, S. EFC/F-BAR proteins and the N-WASP-WIP complex induce membrane curvature-dependent actin polymerization. EMBO J. 27, 2817–2828 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Padrick, S. B. et al. Hierarchical regulation of WASP/WAVE proteins. Mol. Cell 32, 426–438 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Wu, M., Wu, X. & DeCamilli, P. Calcium oscillations-coupled conversion of actin travelling waves to standing oscillations. Proc. Natl Acad. Sci. USA 110, 1339–1344 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Wu, M. et al. Coupling between clathrin-dependent endocytic budding and F-BAR-dependent tubulation in a cell-free system. Nat. Cell Biol. 12, 902–908 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Raucher, D. & Sheetz, M. P. Cell spreading and lamellipodial extension rate is regulated by membrane tension. J. Cell Biol. 148, 127–136 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Sheetz, M. P. Cell control by membrane-cytoskeleton adhesion. Nat. Rev. Mol. Cell Biol. 2, 392–396 (2001).

    Article  CAS  PubMed  Google Scholar 

  28. Diz-Munoz, A. et al. Control of directed cell migration in vivo by membrane-to-cortex attachment. PLoS Biol. 8, e1000544 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Raucher, D. et al. Phosphatidylinositol 4,5-bisphosphate functions as a second messenger that regulates cytoskeleton-plasma membrane adhesion. Cell 100, 221–228 (2000).

    Article  CAS  PubMed  Google Scholar 

  30. Hao, J. J. et al. Phospholipase C-mediated hydrolysis of PIP2 releases ERM proteins from lymphocyte membrane. J. Cell Biol. 184, 451–462 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Ueno, T., Falkenburger, B. H., Pohlmeyer, C. & Inoue, T. Triggering actin comets versus membrane ruffles: distinctive effects of phosphoinositides on actin reorganization. Sci. Signal 4, ra87 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Varnai, P., Thyagarajan, B., Rohacs, T. & Balla, T. Rapidly inducible changes in phosphatidylinositol 4,5-bisphosphate levels influence multiple regulatory functions of the lipid in intact living cells. J. Cell Biol. 175, 377–382 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Itoh, T. et al. Dynamin and the actin cytoskeleton cooperatively regulate plasma membrane invagination by BAR and F-BAR proteins. Dev. Cell 9, 791–804 (2005).

    Article  CAS  PubMed  Google Scholar 

  34. Tsujita, K. et al. Coordination between the actin cytoskeleton and membrane deformation by a novel membrane tubulation domain of PCH proteins is involved in endocytosis. J. Cell Biol. 172, 269–279 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Giannone, G. et al. Periodic lamellipodial contractions correlate with rearward actin waves. Cell 116, 431–443 (2004).

    Article  CAS  PubMed  Google Scholar 

  36. Giannone, G. et al. Lamellipodial actin mechanically links myosin activity with adhesion-site formation. Cell 128, 561–575 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Nolen, B. J. et al. Characterization of two classes of small molecule inhibitors of Arp2/3 complex. Nature 460, 1031–1034 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Gauthier, N. C., Fardin, M. A., Roca-Cusachs, P. & Sheetz, M. P. Temporary increase in plasma membrane tension coordinates the activation of exocytosis and contraction during cell spreading. Proc. Natl Acad. Sci. USA 108, 14467–14472 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Batchelder, E. L. et al. Membrane tension regulates motility by controlling lamellipodium organization. Proc. Natl Acad. Sci. USA 108, 11429–11434 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Fujita, H., Katoh, H., Ishikawa, Y., Mori, K. & Negishi, M. Rapostlin is a novel effector of Rnd2 GTPase inducing neurite branching. J. Biol. Chem. 277, 45428–45434 (2002).

    Article  CAS  PubMed  Google Scholar 

  41. Itoh, T. & DeCamilli, P. BAR, F-BAR (EFC) and ENTH/ANTH domains in the regulation of membrane-cytosol interfaces and membrane curvature. Biochim. Biophys. Acta 1761, 897–912 (2006).

    Article  CAS  PubMed  Google Scholar 

  42. Soderling, S. H. et al. The WRP component of the WAVE-1 complex attenuates Rac-mediated signalling. Nat. Cell Biol. 4, 970–975 (2002).

    Article  CAS  PubMed  Google Scholar 

  43. Guerrier, S. et al. The F-BAR domain of srGAP2 induces membrane protrusions required for neuronal migration and morphogenesis. Cell 138, 990–1004 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Yamazaki, D., Itoh, T., Miki, H. & Takenawa, T. srGAP1 regulates lamellipodial dynamics and cell migratory behavior by modulating Rac1 activity. Mol. Biol. Cell 24, 3393–3405 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Itoh, T., Hasegawa, J., Tsujita, K., Kanaho, Y. & Takenawa, T. The tyrosine kinase Fer is a downstream target of the PLD-PA pathway that regulates cell migration. Sci. Signal. 2, ra52 (2009).

    Article  CAS  PubMed  Google Scholar 

  46. Itoh, T. et al. Dynamin and the actin cytoskeleton cooperatively regulate plasma membrane invagination by BAR and F-BAR proteins. Dev. Cell 9, 791–804 (2005).

    Article  CAS  PubMed  Google Scholar 

  47. Tsujita, K. et al. Coordination between the actin cytoskeleton and membrane deformation by a novel membrane tubulation domain of PCH proteins is involved in endocytosis. J. Cell Biol. 172, 269–279 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Tsujita, K. et al. Antagonistic regulation of F-BAR protein assemblies controls actin polymerization during podosome formation. J. Cell Sci. 126, 2267–2278 (2013).

    Article  CAS  PubMed  Google Scholar 

  49. Shimada, A. et al. Curved EFC/F-BAR-domain dimers are joined end to end into a filament for membrane invagination in endocytosis. Cell 129, 761–772 (2007).

    Article  CAS  PubMed  Google Scholar 

  50. Sheetz, M. P. Cell control by membrane-cytoskeleton adhesion. Nat. Rev. Mol. Cell Biol. 2, 392–396 (2001).

    Article  CAS  PubMed  Google Scholar 

  51. Morlot, S. et al. Membrane shape at the edge of the dynamin helix sets location and duration of the fission reaction. Cell 151, 619–629 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Bhatia, V. K. et al. Amphipathic motifs in BAR domains are essential for membrane curvature sensing. EMBO J. 28, 3303–3314 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We would like to thank T. Inoue for discussions, M. Ichikawa for technical assistance and H. Yamamoto for help with plasmid constructions. This study was supported by a Grant-in-Aid for Scientific Research (C) from the Japan Society for the Promotion of Science (JSPS) to K.T. and T.I., a Grant-in-Aid for Scientific Research (S) from JSPS to T.T. and a Uehara Memorial Foundation grant to K.T.

Author information

Authors and Affiliations

Authors

Contributions

K.T. and T.I. conceived the research project. K.T. and T.I. designed and carried out experiments. T.T. provided key reagents. K.T. and T.I. analysed the data and wrote the manuscript.

Corresponding author

Correspondence to Toshiki Itoh.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 2 PM tension sensitive localisation of FBP17.

(a) Tether force measurement of COS-1 cells. Tether force of identical cell was measured before and after hypotonic shock (upper) or hypertonic shock (lower). Values are presented as ratio relative to before treatment. Mean and s.d. are derived from pooled data from three independent experiments. In total, n = 9 events (hypotonic) and n = 8 events (hypertonic) were analysed, respectively. P = 0.014,P = 0.0012, Student’s t-test. (b) Confocal images of endogenous FBP17, endogenous clathrin and F-actin in COS-1 cell on hypotonic shock. Scale bar, 20 μm. (c) Confocal images of endogenous FBP17 and F-actin (stained with Alexa Fluor 568-conjugated phalloidin) in Swiss 3T3 cells on PDGF stimulation.

Supplementary Figure 3 Manipulation of PM tension affects the localisation of FBP17.

Time-lapse images of COS-1 cell co-expressing GFP-FBP17 and Lifeact-mCherry on addition of deoxycholic acid (400 μM).

Supplementary Figure 4 Models of PM tension fluctuation at the leading edge.

PM at the cell front is pushed outwardly by polymerised actin (blue). When the front membrane retracts backward (a) or curls up by ruffling (b), there emerges a local PM area with reduced tension. The drop in local PM tension induces membrane invaginations by FBP17 (red), which promotes actin assembly. FBP17-mediated actin polymerisation again increases PM tension, resulting in its dissociation from the membrane.

Supplementary Figure 5 Confirmation of effects by RNAi on the expression levels of target proteins.

(a,b) Protein levels in COS-1 cells transfected with the indicated siRNA were analysed by western blot using specific antibodies against FBP17 (a) and N-WASP (b). (c,d) Expressions of Toca family proteins in COS-1 or MDA-MB-231 cells analysed by RT-PCR. Triple RNAi indicates the combination of siRNAs of FBP17, CIP4, and Toca-1.

Supplementary Table 1 Target sequences of siRNA

Supplementary information

Supplementary Information

Supplementary Information (PDF 1323 kb)

Polarisation of FBP17 is induced by PM tension increase.

COS-1cell co-expressing GFP-FBP17 and Lifeact-mCherry was observed by time-lapse microscopy on hypotonic buffer. The movie was taken at 1 frame per 5 s and played at 15 fps. (MOV 14507 kb)

Polarisation of FBP17 is disrupted by PM tension decrease.

COS-1 cell co-expressing GFP-FBP17 and Lifeact-mCherry was observed by time-lapse microscopy on addition of hypertonic buffer. The movie was taken at 1 frame per 5 s and played at 15 fps. (MOV 22709 kb)

Polarisation of FBP17 is induced by PtdIns(4,5)P2 liberation.

COS-1 cell co-expressing GFP-FBP17, CFP-FKBP-PLC δ1 PH domain, and mRFP-FRB-MoA was observed by time-lapse microscopy on addition of rapamycin. The movie was taken at 1 frame per 5 s and played at 15 fps. (MOV 7448 kb)

Polarisation of FBP17 is disrupted by PtdIns(4,5)P2 depletion.

COS-1 cell co-expressing GFP-FBP17, CFP-PM-anchored FRB domain, and mRFP-FKBP-5-phosphatase domain was observed by time-lapse microscopy on addition of rapamycin. The movie was taken at 1 frame per 5 s and played at 15 fps. (MOV 10733 kb)

Dynamics of FBP17 at the leading edge.

COS-1 cell co-expressing GFP-FBP17 and Lifeact-mCherry was observed by time-lapse microscopy. The movie was taken at 1 frame per 5 s and played at 15 fps. (MOV 15287 kb)

Acute disruption of FBP17 polarity by N-WASP inhibition.

COS-1 cell co-expressing GFP-FBP17 and Lifeact-mCherry was observed by time-lapse microscopy on addition of wiskostatin. The movie was taken at 1 frame per 10 s and played at 15 fps. (MOV 3757 kb)

Acute disruption of FBP17 polarity by Arp2/3 complex inhibition.

COS-1 cell co-expressing GFP-FBP17 and Lifeact-mCherry was observed by time-lapse microscopy on addition of CK-666. The movie was taken at 1 frame per 10 s and played at 15 fps. (MOV 8851 kb)

PM tension dependent recruitment of FBP17.

COS-1 cell co-expressing GFP-FBP17 and Lifeact-mCherry was observed by time-lapse microscopy on addition of blebbistatin followed by the treatment of hypertonic buffer. The movie was taken at 1 frame per 5 s and played at 15 fps. (MOV 7257 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Tsujita, K., Takenawa, T. & Itoh, T. Feedback regulation between plasma membrane tension and membrane-bending proteins organizes cell polarity during leading edge formation. Nat Cell Biol 17, 749–758 (2015). https://doi.org/10.1038/ncb3162

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3162

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing